Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
Add more filters

Publication year range
1.
Nat Rev Mol Cell Biol ; 17(7): 426-38, 2016 07.
Article in English | MEDLINE | ID: mdl-27251421

ABSTRACT

RNA helicases comprise the largest family of enzymes involved in the metabolism of mRNAs, the processing and fate of which rely on their packaging into messenger ribonucleoprotein particles (mRNPs). In this Review, we describe how the capacity of some RNA helicases to either remodel or lock the composition of mRNP complexes underlies their pleiotropic functions at different steps of the gene expression process. We illustrate the roles of RNA helicases in coordinating gene expression steps and programmes, and propose that RNA helicases function as molecular drivers and guides of the progression of their mRNA substrates from one RNA-processing factory to another, to a productive mRNA pool that leads to protein synthesis or to unproductive mRNA pools that are stored or degraded.


Subject(s)
Gene Expression Regulation , RNA Helicases/physiology , Animals , Gene Expression , Humans , RNA Splicing , RNA Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism
2.
Genome Res ; 34(3): 394-409, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38508694

ABSTRACT

mRNA translation and decay are tightly interconnected processes both in the context of mRNA quality-control pathways and for the degradation of functional mRNAs. Cotranslational mRNA degradation through codon usage, ribosome collisions, and the recruitment of specific proteins to ribosomes is an important determinant of mRNA turnover. However, the extent to which translation-dependent mRNA decay (TDD) and translation-independent mRNA decay (TID) pathways participate in the degradation of mRNAs has not been studied yet. Here we describe a comprehensive analysis of basal and signal-induced TDD and TID in mouse primary CD4+ T cells. Our results indicate that most cellular transcripts are decayed to some extent in a translation-dependent manner. Our analysis further identifies the length of untranslated regions, the density of ribosomes, and GC3 content as important determinants of TDD magnitude. Consistently, all transcripts that undergo changes in ribosome density within their coding sequence upon T cell activation display a corresponding change in their TDD level. Moreover, we reveal a dynamic modulation in the relationship between GC3 content and TDD upon T cell activation, with a reversal in the impact of GC3- and AU3-rich codons. Altogether, our data show a strong and dynamic interconnection between mRNA translation and decay in mammalian primary cells.


Subject(s)
Lymphocyte Activation , Protein Biosynthesis , RNA Stability , RNA, Messenger , Ribosomes , Ribosomes/metabolism , Animals , Mice , RNA, Messenger/metabolism , RNA, Messenger/genetics , CD4-Positive T-Lymphocytes/metabolism , Mice, Inbred C57BL , T-Lymphocytes/metabolism
3.
Nucleic Acids Res ; 52(4): 1527-1543, 2024 Feb 28.
Article in English | MEDLINE | ID: mdl-38272542

ABSTRACT

The NF-κB protein p65/RelA plays a pivotal role in coordinating gene expression in response to diverse stimuli, including viral infections. At the chromatin level, p65/RelA regulates gene transcription and alternative splicing through promoter enrichment and genomic exon occupancy, respectively. The intricate ways in which p65/RelA simultaneously governs these functions across various genes remain to be fully elucidated. In this study, we employed the HTLV-1 Tax oncoprotein, a potent activator of NF-κB, to investigate its influence on the three-dimensional organization of the genome, a key factor in gene regulation. We discovered that Tax restructures the 3D genomic landscape, bringing together genes based on their regulation and splicing patterns. Notably, we found that the Tax-induced gene-gene contact between the two master genes NFKBIA and RELA is associated with their respective changes in gene expression and alternative splicing. Through dCas9-mediated approaches, we demonstrated that NFKBIA-RELA interaction is required for alternative splicing regulation and is caused by an intragenic enrichment of p65/RelA on RELA. Our findings shed light on new regulatory mechanisms upon HTLV-1 Tax and underscore the integral role of p65/RelA in coordinated regulation of NF-κB-responsive genes at both transcriptional and splicing levels in the context of the 3D genome.


The NF-κB pathway is essential for coordinating gene expression in response to various stimuli, including viral infections. Most studies have focused on the role of NF-κB in transcriptional regulation. In the present study, the impact of the potent NF-κB activator HTLV-1 Tax oncoprotein on the three-dimensional organization of the genome was investigated. Tax-mediated NF-κB activation was found to restructure the 3D genomic landscape in cells and to bring genes together in multigene complexes that are coordinately regulated either transcriptionally or through alternative splicing by NF-κB. Induced coordinate changes in transcription and alternative splicing included the two master genes of NF-κB pathway NFKBIA and RELA. The findings have significant implications for understanding cell fate determination and disease development associated with HTLV-1 infection, as well as chronic NF-κB activation in various human inflammatory diseases and cancer.


Subject(s)
Chromatin Assembly and Disassembly , Gene Expression Regulation , NF-kappa B p50 Subunit , Alternative Splicing/genetics , Chromatin Assembly and Disassembly/genetics , Gene Products, tax/genetics , Gene Products, tax/metabolism , Human T-lymphotropic virus 1/genetics , Human T-lymphotropic virus 1/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Signal Transduction , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , Transcriptional Activation , Humans , NF-kappa B p50 Subunit/metabolism
4.
Nucleic Acids Res ; 50(16): 9226-9246, 2022 09 09.
Article in English | MEDLINE | ID: mdl-36039747

ABSTRACT

DDX5 and DDX17 are DEAD-box RNA helicase paralogs which regulate several aspects of gene expression, especially transcription and splicing, through incompletely understood mechanisms. A transcriptome analysis of DDX5/DDX17-depleted human cells confirmed the large impact of these RNA helicases on splicing and revealed a widespread deregulation of 3' end processing. In silico analyses and experiments in cultured cells showed the binding and functional contribution of the genome organizing factor CTCF to chromatin sites at or near a subset of DDX5/DDX17-dependent exons that are characterized by a high GC content and a high density of RNA Polymerase II. We propose the existence of an RNA helicase-dependent relationship between CTCF and the dynamics of transcription across DNA and/or RNA structured regions, that contributes to the processing of internal and terminal exons. Moreover, local DDX5/DDX17-dependent chromatin loops spatially connect RNA helicase-regulated exons with their cognate promoter, and we provide the first direct evidence that de novo gene looping modifies alternative splicing and polyadenylation. Overall our findings uncover the impact of DDX5/DDX17-dependent chromatin folding on pre-messenger RNA processing.


Subject(s)
DEAD-box RNA Helicases , RNA , Humans , RNA/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , DEAD-box RNA Helicases/metabolism , Alternative Splicing , Chromatin/genetics
5.
Nucleic Acids Res ; 49(18): 10657-10676, 2021 10 11.
Article in English | MEDLINE | ID: mdl-34530456

ABSTRACT

Epstein-Barr virus (EBV) is a human herpesvirus associated with human cancers worldwide. Ex vivo, the virus efficiently infects resting human B lymphocytes and induces their continuous proliferation. This process is accompanied by a global reprogramming of cellular gene transcription. However, very little is known on the impact of EBV infection on the regulation of alternative splicing, a pivotal mechanism that plays an essential role in cell fate determination and is often deregulated in cancer. In this study, we have developed a systematic time-resolved analysis of cellular mRNA splice variant expression during EBV infection of resting B lymphocytes. Our results reveal that major modifications of alternative splice variant expression appear as early as day 1 post-infection and suggest that splicing regulation provides-besides transcription-an additional mechanism of gene expression regulation at the onset of B cell activation and proliferation. We also report a role for the viral proteins, EBNA2 and EBNA-LP, in the modulation of specific alternative splicing events and reveal a previously unknown function for EBNA-LP-together with the RBM4 splicing factor-in the alternative splicing regulation of two important modulators of cell proliferation and apoptosis respectively, NUMB and BCL-X.


Subject(s)
Alternative Splicing , B-Lymphocytes/virology , Herpesvirus 4, Human/physiology , Viral Proteins/metabolism , Cells, Cultured , Exons , Humans , Membrane Proteins/genetics , RNA Splice Sites , RNA-Binding Proteins/metabolism , Viral Proteins/physiology
6.
Genome Res ; 29(5): 711-722, 2019 05.
Article in English | MEDLINE | ID: mdl-30962178

ABSTRACT

The inclusion of exons during the splicing process depends on the binding of splicing factors to short low-complexity regulatory sequences. The relationship between exonic splicing regulatory sequences and coding sequences is still poorly understood. We demonstrate that exons that are coregulated by any given splicing factor share a similar nucleotide composition bias and preferentially code for amino acids with similar physicochemical properties because of the nonrandomness of the genetic code. Indeed, amino acids sharing similar physicochemical properties correspond to codons that have the same nucleotide composition bias. In particular, we uncover that the TRA2A and TRA2B splicing factors that bind to adenine-rich motifs promote the inclusion of adenine-rich exons coding preferentially for hydrophilic amino acids that correspond to adenine-rich codons. SRSF2 that binds guanine/cytosine-rich motifs promotes the inclusion of GC-rich exons coding preferentially for small amino acids, whereas SRSF3 that binds cytosine-rich motifs promotes the inclusion of exons coding preferentially for uncharged amino acids, like serine and threonine that can be phosphorylated. Finally, coregulated exons encoding amino acids with similar physicochemical properties correspond to specific protein features. In conclusion, the regulation of an exon by a splicing factor that relies on the affinity of this factor for specific nucleotide(s) is tightly interconnected with the exon-encoded physicochemical properties. We therefore uncover an unanticipated bidirectional interplay between the splicing regulatory process and its biological functional outcome.


Subject(s)
Alternative Splicing , Exons/genetics , RNA Splice Sites/genetics , RNA Splicing Factors/metabolism , Amino Acids/chemistry , Base Composition/genetics , Cell Line , Genetic Code , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Humans , Introns/genetics , Nucleotide Motifs/genetics , Sequence Analysis, Protein , Sequence Analysis, RNA , Serine-Arginine Splicing Factors/metabolism
7.
Nucleic Acids Res ; 48(5): 2676-2693, 2020 03 18.
Article in English | MEDLINE | ID: mdl-31943118

ABSTRACT

Besides analyses of specific alternative splicing (AS) variants, little is known about AS regulatory pathways and programs involved in anticancer drug resistance. Doxorubicin is widely used in breast cancer chemotherapy. Here, we identified 1723 AS events and 41 splicing factors regulated in a breast cancer cell model of acquired resistance to doxorubicin. An RNAi screen on splicing factors identified the little studied ZRANB2 and SYF2, whose depletion partially reversed doxorubicin resistance. By RNAi and RNA-seq in resistant cells, we found that the AS programs controlled by ZRANB2 and SYF2 were enriched in resistance-associated AS events, and converged on the ECT2 splice variant including exon 5 (ECT2-Ex5+). Both ZRANB2 and SYF2 were found associated with ECT2 pre-messenger RNA, and ECT2-Ex5+ isoform depletion reduced doxorubicin resistance. Following doxorubicin treatment, resistant cells accumulated in S phase, which partially depended on ZRANB2, SYF2 and the ECT2-Ex5+ isoform. Finally, doxorubicin combination with an oligonucleotide inhibiting ECT2-Ex5 inclusion reduced doxorubicin-resistant tumor growth in mouse xenografts, and high ECT2-Ex5 inclusion levels were associated with bad prognosis in breast cancer treated with chemotherapy. Altogether, our data identify AS programs controlled by ZRANB2 and SYF2 and converging on ECT2, that participate to breast cancer cell resistance to doxorubicin.


Subject(s)
Alternative Splicing/genetics , Breast Neoplasms/drug therapy , Doxorubicin/therapeutic use , Drug Resistance, Neoplasm , Proto-Oncogene Proteins/metabolism , RNA-Binding Proteins/metabolism , Adult , Aged , Aged, 80 and over , Alternative Splicing/drug effects , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Exons/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , MCF-7 Cells , Mice, Nude , MicroRNAs/genetics , MicroRNAs/metabolism , Middle Aged , Protein Isoforms/metabolism , RNA Splice Sites/genetics , S Phase/drug effects , Spliceosomes/metabolism
8.
EMBO J ; 35(14): 1565-81, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27266525

ABSTRACT

Condensins associate with DNA and shape mitotic chromosomes. Condensins are enriched nearby highly expressed genes during mitosis, but how this binding is achieved and what features associated with transcription attract condensins remain unclear. Here, we report that condensin accumulates at or in the immediate vicinity of nucleosome-depleted regions during fission yeast mitosis. Two transcriptional coactivators, the Gcn5 histone acetyltransferase and the RSC chromatin-remodelling complex, bind to promoters adjoining condensin-binding sites and locally evict nucleosomes to facilitate condensin binding and allow efficient mitotic chromosome condensation. The function of Gcn5 is closely linked to condensin positioning, since neither the localization of topoisomerase II nor that of the cohesin loader Mis4 is altered in gcn5 mutant cells. We propose that nucleosomes act as a barrier for the initial binding of condensin and that nucleosome-depleted regions formed at highly expressed genes by transcriptional coactivators constitute access points into chromosomes where condensin binds free genomic DNA.


Subject(s)
Adenosine Triphosphatases/metabolism , Chromosomes, Fungal/metabolism , DNA-Binding Proteins/metabolism , Mitosis , Multiprotein Complexes/metabolism , Nucleosomes/metabolism , Schizosaccharomyces/physiology , Acetyltransferases/metabolism , Base Composition , Schizosaccharomyces pombe Proteins/metabolism , Transcription Factors/metabolism
9.
Genome Res ; 27(6): 1087-1097, 2017 06.
Article in English | MEDLINE | ID: mdl-28420690

ABSTRACT

Transcriptomic genome-wide analyses demonstrate massive variation of alternative splicing in many physiological and pathological situations. One major challenge is now to establish the biological contribution of alternative splicing variation in physiological- or pathological-associated cellular phenotypes. Toward this end, we developed a computational approach, named "Exon Ontology," based on terms corresponding to well-characterized protein features organized in an ontology tree. Exon Ontology is conceptually similar to Gene Ontology-based approaches but focuses on exon-encoded protein features instead of gene level functional annotations. Exon Ontology describes the protein features encoded by a selected list of exons and looks for potential Exon Ontology term enrichment. By applying this strategy to exons that are differentially spliced between epithelial and mesenchymal cells and after extensive experimental validation, we demonstrate that Exon Ontology provides support to discover specific protein features regulated by alternative splicing. We also show that Exon Ontology helps to unravel biological processes that depend on suites of coregulated alternative exons, as we uncovered a role of epithelial cell-enriched splicing factors in the AKT signaling pathway and of mesenchymal cell-enriched splicing factors in driving splicing events impacting on autophagy. Freely available on the web, Exon Ontology is the first computational resource that allows getting a quick insight into the protein features encoded by alternative exons and investigating whether coregulated exons contain the same biological information.


Subject(s)
Alternative Splicing , Exons , Gene Expression Profiling/methods , Genome, Human , Molecular Sequence Annotation/methods , Transcriptome , Autophagy , Cell Line, Tumor , Gene Ontology , Genome-Wide Association Study , Humans , MCF-7 Cells , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism , Signal Transduction , Software
10.
Nucleic Acids Res ; 46(15): 7686-7700, 2018 09 06.
Article in English | MEDLINE | ID: mdl-29931089

ABSTRACT

The Repressor Element 1-silencing transcription factor (REST) represses a number of neuronal genes in non-neuronal cells or in undifferentiated neural progenitors. Here, we report that the DEAD box RNA helicase DDX17 controls important REST-related processes that are critical during the early phases of neuronal differentiation. First, DDX17 associates with REST, promotes its binding to the promoter of a subset of REST-targeted genes and co-regulates REST transcriptional repression activity. During neuronal differentiation, we observed a downregulation of DDX17 along with that of the REST complex that contributes to the activation of neuronal genes. Second, DDX17 and its paralog DDX5 regulate the expression of several proneural microRNAs that are known to target the REST complex during neurogenesis, including miR-26a/b that are also direct regulators of DDX17 expression. In this context, we propose a new mechanism by which RNA helicases can control the biogenesis of intronic miRNAs. We show that the processing of the miR-26a2 precursor is dependent on RNA helicases, owing to an intronic regulatory region that negatively impacts on both miRNA processing and splicing of its host intron. Our work places DDX17 in the heart of a pathway involving REST and miRNAs that allows neuronal gene repression.


Subject(s)
DEAD-box RNA Helicases/genetics , Gene Expression Profiling , MicroRNAs/genetics , Repressor Proteins/genetics , Cell Line, Tumor , DEAD-box RNA Helicases/metabolism , Humans , MCF-7 Cells , Neural Stem Cells/metabolism , Neurogenesis/genetics , Neurons/metabolism , Repressor Proteins/metabolism
11.
Bioessays ; 39(10)2017 10.
Article in English | MEDLINE | ID: mdl-28885715

ABSTRACT

The biogenesis of RNAs and proteins is a threat to the cell. Indeed, the act of transcription and nascent RNAs challenge DNA stability. Both RNAs and nascent proteins can also initiate the formation of toxic aggregates because of their physicochemical properties. In reviewing the literature, I show that co-transcriptional and co-translational biophysical constraints can trigger DNA instability that in turn increases the likelihood that sequences that alleviate the constraints emerge over evolutionary time. These directed genetic variations rely on the biogenesis of small RNAs that are transcribed directly from challenged DNA regions or processed from the transcripts that directly or indirectly generate constraints or aggregates. These small RNAs can then target the genomic regions from which they initially originate and increase the local mutation rate of the targeted loci. This mechanism is based on molecular pathways involved in anti-parasite genome defence systems, and implies that gene expression-related biophysical constraints represent a driving force of genome evolution.


Subject(s)
Evolution, Molecular , Genome/genetics , RNA/genetics , Humans , Transcription, Genetic/genetics
12.
Nucleic Acids Res ; 44(W1): W117-21, 2016 07 08.
Article in English | MEDLINE | ID: mdl-27131783

ABSTRACT

Genomic studies and high-throughput experiments often produce large lists of candidate genes among which only a small fraction are truly relevant to the disease, phenotype or biological process of interest. Gene prioritization tackles this problem by ranking candidate genes by profiling candidates across multiple genomic data sources and integrating this heterogeneous information into a global ranking. We describe an extended version of our gene prioritization method, Endeavour, now available for six species and integrating 75 data sources. The performance (Area Under the Curve) of Endeavour on cross-validation benchmarks using 'gold standard' gene sets varies from 88% (for human phenotypes) to 95% (for worm gene function). In addition, we have also validated our approach using a time-stamped benchmark derived from the Human Phenotype Ontology, which provides a setting close to prospective validation. With this benchmark, using 3854 novel gene-phenotype associations, we observe a performance of 82%. Altogether, our results indicate that this extended version of Endeavour efficiently prioritizes candidate genes. The Endeavour web server is freely available at https://endeavour.esat.kuleuven.be/.


Subject(s)
Algorithms , Genetic Predisposition to Disease , Genotype , Software , Animals , Benchmarking , Genetic Association Studies , Humans , Internet , Phenotype
13.
EMBO J ; 32(5): 688-700, 2013 Mar 06.
Article in English | MEDLINE | ID: mdl-23386060

ABSTRACT

Stringent regulation of the interferon (IFN) signalling pathway is essential for maintaining the immune response to pathogens and tumours. The transcription factor STAT1 is a crucial mediator of this response. Here, we show that hCAF1/CNOT7 regulates class I and II IFN pathways at different crucial steps. In resting cells, hCAF1 can control STAT1 trafficking by interacting with the latent form of STAT1 in the cytoplasm. IFN treatment induces STAT1 release, suggesting that hCAF1 may shield cytoplasmic STAT1 from undesirable stimulation. Consistently, hCAF1 silencing enhances STAT1 basal promoter occupancy associated with increased expression of a subset of STAT1-regulated genes. Consequently, hCAF1 knockdown cells exhibit an increased protection against viral infection and reduced viral replication. Furthermore, hCAF1 participates in the extinction of the IFN signal, through its deadenylase activity, by speeding up the degradation of some STAT1-regulated mRNAs. Since abnormal and unbalanced JAK/STAT activation is associated with immune disorders and cancer, hCAF1 could play a major role in innate immunity and oncogenesis, contributing to tumour escape.


Subject(s)
Breast Neoplasms/metabolism , Interferons/pharmacology , STAT1 Transcription Factor/antagonists & inhibitors , Signal Transduction/drug effects , Transcription Factors/metabolism , Virus Replication/drug effects , Apoptosis/drug effects , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Chromatin Immunoprecipitation , Cytoplasm/drug effects , Cytoplasm/metabolism , Exoribonucleases , Female , Fluorescent Antibody Technique , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Immunity, Innate , Immunoprecipitation , MicroRNAs/genetics , Oligonucleotide Array Sequence Analysis , Phosphorylation , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Repressor Proteins , Reverse Transcriptase Polymerase Chain Reaction , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics
14.
Genome Res ; 24(3): 511-21, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24307554

ABSTRACT

Alternative splicing is the main mechanism of increasing the proteome diversity coded by a limited number of genes. It is well established that different tissues or organs express different splicing variants. However, organs are composed of common major cell types, including fibroblasts, epithelial, and endothelial cells. By analyzing large-scale data sets generated by The ENCODE Project Consortium and after extensive RT-PCR validation, we demonstrate that each of the three major cell types expresses a specific splicing program independently of its organ origin. Furthermore, by analyzing splicing factor expression across samples, publicly available splicing factor binding site data sets (CLIP-seq), and exon array data sets after splicing factor depletion, we identified several splicing factors, including ESRP1 and 2, MBNL1, NOVA1, PTBP1, and RBFOX2, that contribute to establishing these cell type-specific splicing programs. All of the analyzed data sets are freely available in a user-friendly web interface named FasterDB, which describes all known splicing variants of human and mouse genes and their splicing patterns across several dozens of normal and cancer cells as well as across tissues. Information regarding splicing factors that potentially contribute to individual exon regulation is also provided via a dedicated CLIP-seq and exon array data visualization interface. To the best of our knowledge, FasterDB is the first database integrating such a variety of large-scale data sets to enable functional genomics analyses at exon-level resolution.


Subject(s)
Alternative Splicing , Epithelial Cells/cytology , Epithelial Cells/metabolism , Fibroblasts/metabolism , RNA-Binding Proteins/metabolism , Animals , Cell Line, Tumor , Exons , Gene Expression Profiling , Human Umbilical Vein Endothelial Cells , Humans , MCF-7 Cells , Mice , Oligonucleotide Array Sequence Analysis , Software , User-Computer Interface
15.
Nucleic Acids Res ; 42(4): 2197-207, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24275493

ABSTRACT

Estrogen and androgen receptors (ER and AR) play key roles in breast and prostate cancers, respectively, where they regulate the transcription of large arrays of genes. The activities of ER and AR are controlled by large networks of protein kinases and transcriptional coregulators, including Ddx5 and its highly related paralog Ddx17. The Ddx5 and Ddx17 RNA helicases are also splicing regulators. Here, we report that Ddx5 and Ddx17 are master regulators of the estrogen- and androgen-signaling pathways by controlling transcription and splicing both upstream and downstream of the receptors. First, Ddx5 and Ddx17 are required downstream of ER and AR for the transcriptional and splicing regulation of a large number of steroid hormone target genes. Second, Ddx5 and Ddx17 act upstream of ER and AR by controlling the expression, at the splicing level, of several key regulators of ER and AR activities. Of particular interest, we demonstrate that Ddx5 and Ddx17 control alternative splicing of the GSK3ß kinase, which impacts on both ER and AR protein stability. We also provide a freely available online resource which gives information regarding splicing variants of genes involved in the estrogen- and androgen-signaling pathways.


Subject(s)
Alternative Splicing , Androgens/pharmacology , DEAD-box RNA Helicases/metabolism , Estrogens/pharmacology , Signal Transduction , Cell Line, Tumor , Dihydrotestosterone/pharmacology , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , MCF-7 Cells , Protein Stability , Receptors, Androgen/metabolism
16.
Nucleic Acids Res ; 41(1): 554-64, 2013 Jan 07.
Article in English | MEDLINE | ID: mdl-23143267

ABSTRACT

It is widely accepted that pre-mRNA maturation, including splicing, is tightly coupled to both transcription and mRNA export, but factors linking the three processes are less understood. By analysing the estrogen-regulated expression of the c-fos mRNA that is processed during transcription, we show that the ddx5 RNA helicase, is required throughout the major nuclear steps of the expression of the c-fos gene, from transcription to mRNA export. Indeed, ddx5, whose recruitment on the c-fos gene was increased upon estrogen treatment, was required for the full transcriptional activation of the c-fos gene. In addition, ddx5 was required for c-fos co-transcriptional RNA splicing. When splicing occurred post-transcriptionally in the absence of ddx5, the c-fos mRNA was poorly exported into the cytosol because of inefficient recruitment of the TAP mRNA export receptor. Finally, ddx5 was present in the c-fos messenger ribonucleoprotein together with mRNA export factors, which further supports that ddx5 is a key operator in the c-fos 'mRNA factory'.


Subject(s)
DEAD-box RNA Helicases/physiology , Proto-Oncogene Proteins c-fos/genetics , RNA, Messenger/metabolism , Transcriptional Activation , Cell Nucleus/metabolism , Estradiol/pharmacology , Humans , MCF-7 Cells , Proto-Oncogene Proteins c-fos/biosynthesis , Proto-Oncogene Proteins c-fos/metabolism , RNA Splicing , RNA Transport , Ribonucleoproteins/metabolism , Transcription, Genetic
17.
Retrovirology ; 11: 119, 2014 Dec 18.
Article in English | MEDLINE | ID: mdl-25519886

ABSTRACT

BACKGROUND: Reprogramming cellular gene transcription sustains HTLV-1 viral persistence that ultimately leads to the development of adult T-cell leukemia/lymphoma (ATLL). We hypothesized that besides these quantitative transcriptional effects, HTLV-1 qualitatively modifies the pattern of cellular gene expression. RESULTS: Exon expression analysis shows that patients' untransformed and malignant HTLV-1(+) CD4(+) T-cells exhibit multiple alternate exon usage (AEU) events. These affect either transcriptionally modified or unmodified genes, culminate in ATLL, and unveil new functional pathways involved in cancer and cell cycle. Unsupervised hierarchical clustering of array data permitted to isolate exon expression patterns of 3977 exons that discriminate uninfected, infected, and transformed CD4(+) T-cells. Furthermore, untransformed infected CD4+ clones and ATLL samples shared 486 exon modifications distributed in 320 genes, thereby indicating a role of AEUs in HTLV-1 leukemogenesis. Exposing cells to splicing modulators revealed that Sudemycin E reduces cell viability of HTLV-1 transformed cells without affecting primary control CD4+ cells and HTLV-1 negative cell lines, suggesting that the huge excess of AEU might provide news targets for treating ATLL. CONCLUSIONS: Taken together, these data reveal that HTLV-1 significantly modifies the structure of cellular transcripts and unmask new putative leukemogenic pathways and possible therapeutic targets.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , Exons , Gene Expression Regulation , Host-Pathogen Interactions , Human T-lymphotropic virus 1/physiology , Leukemia-Lymphoma, Adult T-Cell/pathology , Human T-lymphotropic virus 1/growth & development , Humans , Transcription, Genetic
18.
NAR Cancer ; 6(2): zcae019, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38690580

ABSTRACT

Amino acid bioavailability impacts mRNA translation in a codon-dependent manner. Here, we report that the anti-cancer MAPK inhibitors (MAPKi) decrease the intracellular concentration of aspartate and glutamate in melanoma cells. This coincides with the accumulation of ribosomes on codons corresponding to these amino acids and triggers the translation-dependent degradation of mRNAs encoding aspartate- and glutamate-rich proteins, involved in DNA metabolism such as DNA replication and repair. Consequently, cells that survive MAPKi degrade aspartate and glutamate likely to generate energy, which simultaneously decreases their requirement for amino acids due to the downregulation of aspartate- and glutamate-rich proteins involved in cell proliferation. Concomitantly, the downregulation of aspartate- and glutamate-rich proteins involved in DNA repair increases DNA damage loads. Thus, DNA repair defects, and therefore mutations, are at least in part a secondary effect of the metabolic adaptation of cells exposed to MAPKi.

19.
Database (Oxford) ; 20232023 Dec 21.
Article in English | MEDLINE | ID: mdl-38128543

ABSTRACT

One challenge faced by scientists from the alternative RNA splicing field is to decode the cooperative or antagonistic effects of splicing factors (SFs) to understand and eventually predict splicing outcomes on a genome-wide scale. In this manuscript, we introduce SplicingLore, an open-access database and web resource that help to fill this gap in a straightforward manner. The database contains a collection of RNA-sequencing-derived lists of alternative exons regulated by a total of 75 different SFs. All datasets were processed in a standardized manner, ensuring valid comparisons and correlation analyses. The user can easily retrieve a factor-specific set of differentially included exons from the database or provide a list of exons and search which SF(s) control(s) their inclusion. Our simple workflow is fast and easy to run, and it ensures a reliable calculation of correlation scores between the tested datasets. As a proof of concept, we predicted and experimentally validated a novel functional cooperation between the RNA helicases DDX17 and DDX5 and the heterogeneous nuclear ribonucleoprotein C (HNRNPC) protein. SplicingLore is available at https://splicinglore.ens-lyon.fr/. Database URL:  https://splicinglore.ens-lyon.fr/.


Subject(s)
Alternative Splicing , RNA Splicing , Humans , RNA Splicing Factors/genetics , RNA Splicing/genetics , Genome , Exons/genetics
20.
Nucleic Acids Res ; 38(9): 2825-38, 2010 May.
Article in English | MEDLINE | ID: mdl-20110256

ABSTRACT

It has been shown that alternative splicing is especially prevalent in brain and testis when compared to other tissues. To test whether there is a specific propensity of these tissues to generate splicing variants, we used a single source of high-density microarray data to perform both splicing factor and exon expression profiling across 11 normal human tissues. Paired comparisons between tissues and an original exon-based statistical group analysis demonstrated after extensive RT-PCR validation that the cerebellum, testis, and spleen had the largest proportion of differentially expressed alternative exons. Variations at the exon level correlated with a larger number of splicing factors being expressed at a high level in the cerebellum, testis and spleen than in other tissues. However, this splicing factor expression profile was similar to a more global gene expression pattern as a larger number of genes had a high expression level in the cerebellum, testis and spleen. In addition to providing a unique resource on expression profiling of alternative splicing variants and splicing factors across human tissues, this study demonstrates that the higher prevalence of alternative splicing in a subset of tissues originates from the larger number of genes, including splicing factors, being expressed than in other tissues.


Subject(s)
Alternative Splicing , RNA-Binding Proteins/metabolism , Exons , Gene Expression Profiling , Humans , Oligonucleotide Array Sequence Analysis , RNA-Binding Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL